Open Full Page

Total Page:16

File Type:pdf, Size:1020Kb

Open Full Page Published OnlineFirst May 4, 2016; DOI: 10.1158/1078-0432.CCR-15-2709 CCR Drug Updates Clinical Cancer Research Talimogene Laherparepvec for the Treatment of Advanced Melanoma Patrick A. Ott1,2 and F. Stephen Hodi1,2 Abstract Talimogene laherparepvec (T-VEC) is a first-in-class oncolytic skin or lymph nodes. The drug is currently in clinical trials as virus that mediates local and systemic antitumor activity by direct monotherapy and in combination with immune-checkpoint inhi- cancer cell lysis and an "in situ vaccine" effect. Based on an increased bitors and radiotherapy in melanoma and other cancers. The durable response rate compared with granulocyte macrophage– mechanism of action, toxicity, and efficacy as well as its role in colony stimulating factor in a randomized phase III trial, it was current clinical practice and potential future applications are approved by the FDA for the treatment of melanoma metastatic to reviewed. Clin Cancer Res; 22(13); 3127–31. Ó2016 AACR. Introduction factor (GM-CSF) in the process. The genes encoding neuroviru- lence infected cell protein 34.5 (ICP34.5) and the infected cell Novel systemic treatment modalities such as inhibition of the protein 47 (ICP47) are functionally deleted in the virus, while the immune checkpoints CTLA-4 and PD-1/PD-L1 as well as BRAF gene for human GM-CSF is inserted. ICP34.5 is required for viral and MEK inhibition have expanded the range of therapeutic replication in normal cells, which is mediated by interaction with modalities for advanced melanoma (1–11). The antitumor activ- proliferating cell nuclear antigen (PCNA; ref. 14), whereas cancer ity of both MAPK pathway–targeted therapy (for BRAFV600- cells proliferate independently of ICP34.5 expression. ICP47 is mutant melanoma) and immune checkpoint inhibition (inde- critical for the evasion of HSV-infected cells from cytotoxic T cells pendent of a BRAF mutation) with response rates of 60% and by interfering with peptide processing and presentation on higher is striking and has improved the prognosis for many MHC-1 (15). Deletion of ICP47 in T-VEC prevents potentially patients. Both CTLA-4 and/or PD-1/PD-L1 blockade with mono- limited viral antigen presentation, which could compromise its clonal antibodies can achieve durable clinical benefit, suggesting function as an in situ vaccine. ICP47 deletion also leads to that endogenous tumor directed T-cell responses, suppressed by increased expression of the US11 gene, resulting in increased inhibitory pathways such as CTLA-4 and/or PD-1/PD-L1, can be virus replication in cancer cells without decreasing tumor selec- invigorated, resulting in effective tumor control (1, 2, 10–13). tivity. GM-CSF is a proinflammatory cytokine that promotes the Many patients experience primary or secondary resistance to PD-1 recruitment and maturation of dendritic cells (DC) as well as and/or CTLA-4 inhibition. Alternative treatments for these macrophages into potent antigen-presenting cells, leading to patients are therefore still urgently needed. Talimogene laherpar- priming of tumor-specific T cells (16). It has been used success- epvec (T-VEC), an agent with a different and potentially comple- fully as an immune adjuvant in many cancer vaccines. mentary mechanism of action to immune checkpoint blockade, is T-VEC has two distinct mechanisms of action: The lytic a recent addition to the therapeutic armamentarium for patients function of the virus destroys tumor cells directly, whereas the with advanced melanoma. lysis of the cancer cells leads to release of tumor antigens, virus, and GM-CSF, attracting DCs, thereby creating an in situ vaccine Mechanism of Action (Fig. 1). In a subcutaneous murine melanoma model, tumor T-VEC is an intralesionally delivered oncolytic immunotherapy growth inhibition on the contralateral, uninjected site was only comprised of a genetically engineered attenuated herpes simplex seen when T-VEC contained GM-CSF, establishing a systemic virus type 1 (HSV-1) of the JS-1 strain. T-VEC invades both effect of the lytic virus that is likely mediated by a host immune cancerous and healthy cells but can only replicate in cancer cells, response (17, 18). where it secretes granulocyte macrophage–colony stimulating Clinical Development Phase I 1Department of Medical Oncology, Melanoma Disease Center, and Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard In a phase I study, 30 patients with previously treated mela- Medical School, Boston, Massachusetts. 2Department of Medicine, noma, breast cancer, gastric adenocarcinoma, or head and neck Brigham and Women's Hospital, Boston, Massachusetts. cancer who had cutaneous or subcutaneous lesions accessible Corresponding Author: Patrick A. Ott, Melanoma Disease Center & Center for for injections were treated with different doses and schedules Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Bos- of T-VEC (19). The most common adverse events were grade 1 ton, MA 02215. Phone: 617-582-9030; Fax: 617-632-6727; E-mail: fever, constitutional symptoms, nausea, anorexia, and injec- [email protected] tion site reactions. One patient was reported to experience doi: 10.1158/1078-0432.CCR-15-2709 grade 2 fever, rigor, hypotension, tachycardia, and constitu- Ó2016 American Association for Cancer Research. tional symptoms. Overall, the toxicities were more intense in www.aacrjournals.org 3127 Downloaded from clincancerres.aacrjournals.org on October 3, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst May 4, 2016; DOI: 10.1158/1078-0432.CCR-15-2709 Ott and Hodi Dendritic cells Figure 1. In situ vaccination effect of T-VEC, potentially converting a non–T-cell- inflamed tumor to a T-cell–inflamed T-VEC Lysed tumor tumor. Replication of T-VEC in tumor cells cells leads to their lysis with release of tumor antigens, viral antigens and pathogen-associated molecular Inflammatory patterns (viral DNA, RNA, and cytokines T cells proteins), and GM-CSF. This process (IFNγ, TNFβ) results in the recruitment and Noninflamed GM-CSF Inflamed maturation of antigen-presenting cells, “cold” tumor “hot” tumor including dendritic cells, which present tumor antigens to cytotoxic CD8 T cells. © 2016 American Association for Cancer Research HSV-seronegative patients; an initial low dose of T-VEC, lead- was administered until CR, clinically significant progressive dis- ing to HSV seroconversion, followed by a series of higher-dose ease, intolerable side effects, or 12 months of therapy without an injections was better tolerated. There were no partial or com- objective response. As in the phase II study, T-VEC was initially plete responses (CR); however, flattening of both injected administered at 106 pfu/mL for seroconversion, whereas subse- and noninjected metastases was seen in 6 of 26 evaluable quent doses were given at 108 pfu/mL 3 weeks after the first dose patients. Posttreatment biopsies of injected lesions showed and then every 2 weeks. T-VEC injection was restricted to cuta- inflammation and necrosis. neous and subcutaneous metastases; different lesions could be prioritized for injection differently at any visit depending on its size and the emergence of new lesions. GM-CSF was given daily Phase II subcutaneously at 125 mg/m2 during the first 14 days of a 28-day Fifty patients with unresectable stage IIIC–IV melanoma with cycle; it was chosen as a comparator arm based on overall survival one or more injection-accessible tumor lesions were enrolled (OS) benefit compared with historical controls observed in a in a phase II study assessing the response rate, survival, and safety previous study in melanoma patients at high risk for recurrence of T-VEC (20). Thirty-seven (74%) of the patients had received (23). The primary endpoint was durable response rate (DRR), prior systemic therapy and 20 (40%) had M1c visceral disease. defined as PR or CR with an onset during the first 12 months of Based on the experience from the phase I study, patients received treatment and lasting for at least 6 months. Secondary endpoints intratumoral injections of up to 4 mL of 106 pfu/mL of T-VEC, included OS, best overall response, and duration of response. followed 3 weeks later by up to 4 mL of 108 pfu/mL, and Approximately half of the patients in each arm were previously subsequently every 2 weeks for a maximum of 24 treatments. In untreated; 45% of patients in the T-VEC arm and 39% of patients a small subset of patients, peripheral blood and tumor biopsies þ þ in the GM-CSF arm were stage IVM1b/c. The study met its primary were obtained for assessment of effector T cells, CD4 FoxP3 þ þ endpoint: DRR was significantly higher in the T-VEC arm (16.3%) regulatory T cells (Treg), CD8 FoxP3 suppressor T cells, and compared with the GM-CSF arm (2.1%). The overall response rate myeloid-derived suppressive cells (MDSC; ref. 21). Eight CRs and was also significantly increased in the T-VEC arm (26.4%) com- five partial responses (PR) were observed, resulting in an overall pared with GM-CSF alone (5.7%) as was the number of CRs response rate (ORR) of 26%. Twelve of the 13 responses lasted (10.8% vs. 1%). The median OS was 23.3 months in the T-VEC longer than 6 months. Compared with untreated melanoma arm and 18.9 months in the GM-CSF arm (HR, 0.79; 95% CI, lesions, melanoma metastases regressing after treatment with 0.62–1.00; P ¼ 0.051), and it was therefore unclear, at least from T-VEC exhibited an increase of MART-1–specific T cells compared the primary study analysis, whether T-VEC was associated with with melanoma lesions from untreated patients, whereas num- improved OS. bers of Tregs and MDSCs were decreased. Evidence for increased Subgroup analyses showed higher antitumor activity of T-VEC MART-1–specific T cells was seen in tumor-infiltrating lympho- in patients with stage IIIB, IIIC, and IVM1a disease: with T-VEC, cytes (TIL) and peripheral blood from a patient with a CR after T- DRR was 33% in patients with IIIB or IIIC and 16% in patients TVEC, suggesting the induction of a systemic melanoma-specific with stage IVM1a, respectively compared with 0% and 2% with immune response.
Recommended publications
  • Pharmacological Improvement of Oncolytic Virotherapy
    PHARMACOLOGICAL IMPROVEMENT OF ONCOLYTIC VIROTHERAPY Mohammed Selman Thesis submitted to the Faculty of Graduate and Postdoctoral Studies in partial fulfillment of the requirements for the Doctorate of Philosophy in Biochemistry Department of Biochemistry, Microbiology & Immunology Faculty of Medicine University of Ottawa © Mohammed Selman, Ottawa, Canada, 2018 Abstract Oncolytic viruses (OV) are an emerging class of anticancer bio-therapeutics that induce antitumor immunity through selective replication in cancer cells. However, the efficacy of OVs as single agents remains limited. We postulate that resistance to oncolytic virotherapy results in part from the failure of tumor cells to be sufficiently infected. In this study, we provide evidence that in the context of sarcoma, a highly heterogeneous malignancy, the infection of tumors by different oncolytic viruses varies greatly. Similarly, for a given oncolytic virus, productive infection of tumors across patient samples varies by many orders of magnitude. To overcome this issue, we hypothesize that the infection of resistant tumors can be achieved through the use of selected small molecules. Here, we have identified two novel drug classes with the ability to improve the efficacy of OV therapy: fumaric and maleic acid esters (FMAEs) and vanadium compounds. FMAEs are enhancing infection of cancer cells by several oncolytic viruses in cancer cell lines and human tumor biopsies. The ability of FMAEs to enhance viral spread is due to their ability to inhibit type I IFN production and response, which is associated with their ability to block nuclear translocation of transcription factor NF-κB. Vanadium-based phosphatase inhibitors enhance OV infection of RNA viruses in vitro and ex vivo, in resistant cancer cell lines.
    [Show full text]
  • (T-VEC): an Intralesional Cancer Immunotherapy for Advanced Melanoma
    cancers Review Talimogene Laherparepvec (T-VEC): An Intralesional Cancer Immunotherapy for Advanced Melanoma Pier Francesco Ferrucci 1,* , Laura Pala 2, Fabio Conforti 2 and Emilia Cocorocchio 3 1 Tumor Biotherapy Unit, Department of Experimental Oncology, European Institute of Oncology, IRCCS, 20141 Milan, Italy 2 Division of Melanoma, Sarcoma and Rare Tumors, European Institute of Oncology, IRCCS, 20141 Milan, Italy; [email protected] (L.P.); [email protected] (F.C.) 3 Hemato-Oncology Division, European Institute of Oncology, IRCCS, 20141 Milan, Italy; [email protected] * Correspondence: [email protected]; Tel.: +39-0294371094 Simple Summary: Talimogene laherparepvec (T-VEC; IMLYGIC®, Amgen Inc.) is the first oncolytic vi- ral immunotherapy to be approved for the local treatment of unresectable metastatic stage IIIB/C–IVM1a melanoma. Its direct intratumoral injection aim to trigger local and systemic immunologic responses leading to tumor cell lysis, followed by release of tumor-derived antigens and subsequent activation of tumor-specific effector T-cells. Its approval has fueled the interest to study its possible sinergy with other immunotherapeutics in preclinical models as well as in clinical contextes. In fact, it has been shown that intratumoral administration of this immunostimulatory agent successfully synergizes with immune checkpoint inhibitors. The objectives of this review are to resume the current state of the art of T-VEC treatment when used in monotherapy or in combination with immune checkpoint inhibitors, describing the strong rationale of its development, the Citation: Ferrucci, P.F.; Pala, L.; adverse events of interest and the clinical outcome in selected patient’s populations. Conforti, F.; Cocorocchio, E.
    [Show full text]
  • Intratumoral Injection of HSV1716, an Oncolytic Herpes Virus, Is Safe and Shows Evidence of Immune Response and Viral Replication in Young Cancer Patients Keri A
    Published OnlineFirst May 11, 2017; DOI: 10.1158/1078-0432.CCR-16-2900 Cancer Therapy: Clinical Clinical Cancer Research Intratumoral Injection of HSV1716, an Oncolytic Herpes Virus, Is Safe and Shows Evidence of Immune Response and Viral Replication in Young Cancer Patients Keri A. Streby1,2, James I. Geller3, Mark A. Currier2, Patrick S. Warren4, John M. Racadio5, Alexander J. Towbin5, Michele R. Vaughan1, Melinda Triplet1, Kristy Ott-Napier1, Devon J. Dishman1, Lori R. Backus3, Beth Stockman3, Marianne Brunner6, Kathleen Simpson7, Robert Spavin7, Joe Conner7, and Timothy P. Cripe1,2 Abstract Purpose: HSV1716 is an oncolytic herpes simplex virus-1 included low-grade fever, chills, and mild cytopenias. Six of (HSV-1) studied in adults via injection into the brain and super- eight HSV-1 seronegative patients at baseline showed serocon- ficial tumors. To determine the safety of administering HSV1716 version on day 28. Six of nine patients had detectable HSV-1 to pediatric patients with cancer, we conducted a phase I trial of genomes by PCR in peripheral blood appearing on day þ4 image-guided injection in young patients with relapsed or refrac- consistent with de novo virus replication. Two patients had tory extracranial cancers. transient focal increases in metabolic activity on 18fluorine- Experimental Design: We delivered a single dose of 105 to 107 deoxyglucose PET, consistent with inflammatory reactions. In infectious units of HSV1716 via computed tomography–guided onecase,thesamegeographicregionthatflared later appeared intratumoral injection and measured tumor responses by imag- necrotic on imaging. No patient had an objective response to ing. Patients were eligible for up to three more doses if they HSV1716.
    [Show full text]
  • Oncolytic Virotherapy with an HSV Amplicon Vector Expressing
    Cancer Gene Therapy (2007) 14, 918–926 r 2007 Nature Publishing Group All rights reserved 0929-1903/07 $30.00 www.nature.com/cgt ORIGINAL ARTICLE Oncolytic virotherapy with an HSV amplicon vector expressing granulocyte–macrophage colony-stimulating factor using the replication-competent HSV type 1 mutant HF10 as a helper virus S-i Kohno1,2, C Luo1,2, A Nawa3, Y Fujimoto4, D Watanabe5, F Goshima1, T Tsurumi6 and Y Nishiyama1 1Department of Virology, Graduate School of Medicine, Nagoya University, Nagoya, Japan; 2Research Division, M’s Science Corporation, Kobe, Japan; 3Department of Obstetrics and Genecology, Graduate School of Medicine, Nagoya University, Nagoya, Japan; 4Department of Otorhinolaryngology, Graduate School of Medicine, Nagoya University, Nagoya, Japan; 5Department of Dermatology, Aichi Medical University, Aichi, Japan and 6Division of Virology, Aichi Cancer Center Research Institute, Nagoya, Japan Direct viral infection of solid tumors can cause tumor cell death, but these techniques offer the opportunity to express exogenous factors to enhance the antitumor response. We investigated the antitumor effects of a herpes simplex virus (HSV) amplicon expressing mouse granulocyte–macrophage colony-stimulating factor (mGM-CSF) using the replication-competent HSV type 1 mutant HF10 as a helper virus. HF10-packaged mGM-CSF-expressing amplicon (mGM-CSF amplicon) was used to infect subcutaneously inoculated murine colorectal tumor cells (CT26 cells) and the antitumor effects were compared to tumors treated with only HF10. The mGM-CSF amplicon efficiently replicated in CT26 cells with similar oncolytic activity to HF10 in vitro. However, when mice subcutaneously inoculated with CT26 cells were intratumorally injected with HF10 or mGM-CSF amplicon, greater tumor regression was seen in mGM-CSF amplicon-treated animals.
    [Show full text]
  • Oncolytic Herpes Simplex Virus-Based Therapies for Cancer
    cells Review Oncolytic Herpes Simplex Virus-Based Therapies for Cancer Norah Aldrak 1 , Sarah Alsaab 1,2, Aliyah Algethami 1, Deepak Bhere 3,4,5 , Hiroaki Wakimoto 3,4,5,6 , Khalid Shah 3,4,5,7, Mohammad N. Alomary 1,2,* and Nada Zaidan 1,2,* 1 Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11451, Saudi Arabia; [email protected] (N.A.); [email protected] (S.A.); [email protected] (A.A.) 2 National Center for Biotechnology, Life Science and Environmental Research Institute, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11451, Saudi Arabia 3 Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; [email protected] (D.B.); [email protected] (H.W.); [email protected] (K.S.) 4 Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA 5 BWH Center of Excellence for Biomedicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA 6 Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA 7 Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA * Correspondence: [email protected] (M.N.A.); [email protected] (N.Z.) Abstract: With the increased worldwide burden of cancer, including aggressive and resistant cancers, oncolytic virotherapy has emerged as a viable therapeutic option. Oncolytic herpes simplex virus (oHSV) can be genetically engineered to target cancer cells while sparing normal cells.
    [Show full text]
  • Oncolytic Virotherapy Blockade by Microglia and Macrophages Requires STAT1/3 Zahid M
    Published OnlineFirst November 8, 2017; DOI: 10.1158/0008-5472.CAN-17-0599 Cancer Translational Science Research Oncolytic Virotherapy Blockade by Microglia and Macrophages Requires STAT1/3 Zahid M. Delwar1,2,3, Yvonne Kuo1, Yan H. Wen1,4, Paul S. Rennie3, and William Jia1,2 Abstract The first oncolytic virotherapy employing HSV-1 (oHSV-1) STAT1/3 was determined to be responsible for suppressing was approved recently by the FDA to treat cancer, but further oHSV-1 replication in macrophages/microglia. Treatment improvements in efficacy are needed to eradicate challenging with the oxindole/imidazole derivative C16 rescued oHSV-1 refractory tumors, such as glioblastomas (GBM). Microglia/ replication in microglia/macrophages by inhibiting STAT1/3 macrophages comprising approximately 40% of a GBM tumor activity. In the U87 xenograft model of GBM, C16 treatment may limit virotherapeutic efficacy. Here, we show these cells overcame the microglia/macrophage barrier, thereby facilitat- suppress oHSV-1 growth in gliomas by internalizing the virus ing tumor regression without causing a spread of the virus to through phagocytosis. Internalized virus remained capable of normal organs. Collectively, our results suggest a strategy to expressing reporter genes while viral replication was blocked. relieve a STAT1/3-dependent therapeutic barrier and enhance Macrophage/microglia formed a nonpermissive OV barrier, oHSV-1 oncolytic activity in GBM. preventing dissemination of oHSV-1 in the glioma mass. The Significance: These findings suggest a strategy to enhance the deficiency in viral replication in microglial cells was associated therapeutic efficacy of oncolytic virotherapy in glioblastoma. with silencing of particular viral genes. Phosphorylation of Cancer Res; 78(3); 718–30.
    [Show full text]
  • Intraarterial Delivery of Virotherapy for Glioblastoma
    NEUROSURGICAL FOCUS Neurosurg Focus 50 (2):E7, 2021 Intraarterial delivery of virotherapy for glioblastoma Visish M. Srinivasan, MD,1 Frederick F. Lang, MD,2 and Peter Kan, MD3 1Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona; 2Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas; and 3Department of Neurosurgery, University of Texas Medical Branch, Galveston, Texas Oncolytic viruses (OVs) have been used in the treatment of cancer, in a focused manner, since the 1990s. These OVs have become popular in the treatment of several cancers but are only now gaining interest in the treatment of glioblas- toma (GBM) in recent clinical trials. In this review, the authors discuss the unique applications of intraarterial (IA) delivery of OVs, starting with concepts of OV, how they apply to IA delivery, and concluding with discussion of the current ongo- ing trials. Several OVs have been used in the treatment of GBM, including specifically several modified adenoviruses. IA delivery of OVs has been performed in the hepatic circulation and is now being studied in the cerebral circulation to help enhance delivery and specificity. There are some interesting synergies with immunotherapy and IA delivery of OVs. Some of the shortcomings are discussed, specifically the systemic response to OVs and feasibility of treatment. Future studies can be performed in the preclinical setting to identify the ideal candidates for translation into clinical trials, as well as the nuances of this novel delivery method. https://thejns.org/doi/abs/10.3171/2020.11.FOCUS20845 KEYWORDS endovascular; intraarterial; glioblastoma; virotherapy; oncolytic; stem cell NCOLYTIC viruses (OVs) have been tested since the Intraarterial (IA) delivery of OVs has been established in 1950s;1–3 however, it was not until the 1990s1 that other cancers and has had early applications in GBM as viral genomic engineering resulted in the first gen- well.
    [Show full text]
  • T-VEC, Formerly Oncovexgm-CSF), Which Is Being Developed by CTEP in Collaboration with Amgen
    REQUEST FOR PROJECT TEAM MEMBER APPLICATION FOR CONDUCTING CLINICAL TRIALS USING TALIMOGENE LAHERPAREPVEC The Cancer Therapy Evaluation Program (CTEP) is accepting Project Team Member Applications (PTMAs) for a project using talimogene laherparepvec (T-VEC, formerly OncoVEXGM-CSF), which is being developed by CTEP in collaboration with Amgen. T-VEC is a herpes simplex virus type 1 (HSV-1) genetically engineered to selectively replicate in tumor cells and produce human granulocyte-macrophage colony-stimulating factor (GM-CSF) (Liu et al., 2003). T-VEC is the first oncolytic immunotherapy to demonstrate clinical benefit in patients with melanoma in a phase 3 clinical study (Lichty et al., 2014; Andtbacka et al., 2015a). Solicitation of the Project Team Member Application: At the present time, the initial CTEP development plan will include two to three clinical trials, focusing on the objectives of understanding the mechanisms of action and expanding the therapeutic potential of T-VEC. The clinical settings being considered include locally advanced/metastatic breast cancer, bladder cancer, colorectal cancer, sarcomas, non-melanoma skin cancers, or other indications where intralesional injections are feasible. Investigational regimens may be based on monotherapy or combination with immune modulators such as PD-1 or CTLA-4 antagonists, other immunotherapies, or tumor-targeted therapies including standard of care. CTEP invites the investigators to apply in the following categories: 1. Clinician-scientists with expertise in clinical studies of oncolytic virus, cancer immunotherapy, or the select clinical setting. (fill out Part A of the attached Application); 2. Translational scientists with expertise in biomarker studies, especially assessment of immunogenic cell death and cancer immune monitoring.
    [Show full text]
  • The Current Landscape of Oncolytic Herpes Simplex Viruses As Novel Therapies for Brain Malignancies
    viruses Review The Current Landscape of Oncolytic Herpes Simplex Viruses as Novel Therapies for Brain Malignancies Joshua D. Bernstock 1,† , Samantha E. Hoffman 1,2,†, Jason A. Chen 1, Saksham Gupta 1, Ari D. Kappel 1, Timothy R. Smith 1,3 and E. Antonio Chiocca 1,* 1 Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; [email protected] (J.D.B.); [email protected] (S.E.H.); [email protected] (J.A.C.); [email protected] (S.G.); [email protected] (A.D.K.); [email protected] (T.R.S.) 2 Harvard-MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA 3 Computational Neuroscience Outcomes Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA * Correspondence: [email protected] † These authors contributed equally. Abstract: Despite advances in surgical resection and chemoradiation, high-grade brain tumors continue to be associated with significant morbidity/mortality. Novel therapeutic strategies and approaches are, therefore, desperately needed for patients and their families. Given the success experienced in treating multiple other forms of cancer, immunotherapy and, in particular, im- munovirotherapy are at the forefront amongst novel therapeutic strategies that are currently under investigation for incurable brain tumors. Accordingly, herein, we provide a focused mini review of pertinent oncolytic herpes viruses (oHSV) that are being investigated in clinical trials. Citation: Bernstock, J.D.; Hoffman, S.E.; Chen, J.A.; Gupta, S.; Kappel, Keywords: oncolytic virus; herpes simplex virus (HSV); immunotherapy; immunovirotherapy; A.D.; Smith, T.R.; Chiocca, E.A. The glioblastoma (GBM); brain tumors Current Landscape of Oncolytic Herpes Simplex Viruses as Novel Therapies for Brain Malignancies.
    [Show full text]
  • Intratumoral OH2, an Oncolytic Herpes Simplex Virus 2, in Patients with Advanced Solid Tumors: a Multicenter, Phase I/II Clinical Trial
    Open access Original research J Immunother Cancer: first published as 10.1136/jitc-2020-002224 on 9 April 2021. Downloaded from Intratumoral OH2, an oncolytic herpes simplex virus 2, in patients with advanced solid tumors: a multicenter, phase I/II clinical trial 1 1,2 1 3 4 5 Bo Zhang, Jing Huang , Jialin Tang, Sheng Hu, Suxia Luo, Zhiguo Luo, Fuxiang Zhou,6 Shiyun Tan,7 Jieer Ying,8 Qing Chang,9 Rui Zhang,9 Chengyun Geng,9 Dawei Wu,10 Xiangyong Gu,11 Binlei Liu11,12 To cite: Zhang B, Huang J, ABSTRACT with immune checkpoint inhibitors in selected tumor types Tang J, et al. Intratumoral Background OH2 is a genetically engineered oncolytic is warranted. OH2, an oncolytic herpes herpes simplex virus type 2 designed to selectively amplify simplex virus 2, in patients in tumor cells and express granulocyte- macrophage with advanced solid tumors: a colony- stimulating factor to enhance antitumor immune BACKGROUND multicenter, phase I/II clinical Oncolytic virotherapy represents a unique trial. Journal for ImmunoTherapy responses. We investigated the safety, tolerability and of Cancer 2021;9:e002224. antitumor activity of OH2 as single agent or in combination antitumor strategy using natural or geneti- doi:10.1136/jitc-2020-002224 with HX008, an anti- programmed cell death protein 1 cally engineered viruses to infect and repli- antibody, in patients with advanced solid tumors. cate in tumor cells. The mechanisms of the ► Additional supplemental Methods In this multicenter, phase I/II trial, we enrolled tumoricidal effect include direct tumor cell material is published online only. patients with standard treatment- refractory advanced solid lysis caused by selective infection, and the To view, please visit the journal tumors who have injectable lesions.
    [Show full text]
  • HSV-1 Oncolytic Viruses from Bench to Bedside: an Overview of Current Clinical Trials
    cancers Review HSV-1 Oncolytic Viruses from Bench to Bedside: An Overview of Current Clinical Trials Marilin S. Koch, Sean E. Lawler * and E. Antonio Chiocca Harvey Cushing Neurooncology Research Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; [email protected] (E.A.C.) * Correspondence: [email protected] Received: 22 October 2020; Accepted: 23 November 2020; Published: 26 November 2020 Simple Summary: Oncolytic Herpes simplex virus-1 (HSV-1) offers the dual potential of both lytic tumor-specific cell killing and inducing anti-tumor immune responses. The HSV-1 genome can be altered to enhance both components and this may be applicable for the treatment of a broad range of cancers. Several engineered oncolytic viruses based on the HSV-1 backbone are currently under investigation in various clinical trials, both as single agents and in combination with various immunomodulatory drugs. Abstract: Herpes simplex virus 1 (HSV-1) provides a genetic chassis for several oncolytic viruses (OVs) currently in clinical trials. Oncolytic HSV1 (oHSV) have been engineered to reduce neurovirulence and enhance anti-tumor lytic activity and immunogenicity to make them attractive candidates in a range of oncology indications. Successful clinical data resulted in the FDA-approval of the oHSV talimogene laherparepvec (T-Vec) in 2015, and several other variants are currently undergoing clinical assessment and may expand the landscape of future oncologic therapy options. This review offers a detailed overview of the latest results from clinical trials as well as an outlook on newly developed HSV-1 oncolytic variants with improved tumor selectivity, replication, and immunostimulatory capacity and related clinical studies.
    [Show full text]
  • Talimogene Laherparepvec Improves Durable Response Rate in Patients with Advanced Melanoma Robert H.I
    VOLUME 33 ⅐ NUMBER 25 ⅐ SEPTEMBER 1 2015 JOURNAL OF CLINICAL ONCOLOGY ORIGINAL REPORT Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma Robert H.I. Andtbacka, Howard L. Kaufman, Frances Collichio, Thomas Amatruda, Neil Senzer, Jason Chesney, Keith A. Delman, Lynn E. Spitler, Igor Puzanov, Sanjiv S. Agarwala, Mohammed Milhem, Lee Cranmer, Brendan Curti, Karl Lewis, Merrick Ross, Troy Guthrie, Gerald P. Linette, Gregory A. Daniels, Kevin Harrington, Mark R. Middleton, Wilson H. Miller Jr, Jonathan S. Zager, Yining Ye, Bin Yao, Ai Li, Susan Doleman, Ari VanderWalde, Jennifer Gansert, and Robert S. Coffin See accompanying article on page 2812 Author affiliations appear at the end of this article. ABSTRACT Published online ahead of print at Purpose www.jco.org on June 22, 2015. Talimogene laherparepvec (T-VEC) is a herpes simplex virus type 1–derived oncolytic immuno- Written on behalf of the OPTiM therapy designed to selectively replicate within tumors and produce granulocyte macrophage investigators. colony-stimulating factor (GM-CSF) to enhance systemic antitumor immune responses. T-VEC Supported by Amgen, which also was compared with GM-CSF in patients with unresected stage IIIB to IV melanoma in a funded medical writing assistance. randomized open-label phase III trial. R.H.I.A. and H.L.K. contributed equally to this work. Patients and Methods Patients with injectable melanoma that was not surgically resectable were randomly assigned at Presented in part at the 49th Annual a two-to-one ratio to intralesional T-VEC or subcutaneous GM-CSF. The primary end point was Meeting of the American Society of durable response rate (DRR; objective response lasting continuously Ն 6 months) per independent Clinical Oncology, Chicago, IL, May 31-June 4, 2013, and 50th ASCO assessment.
    [Show full text]